1
|
Goyal S, Tibrewal S, Ratna R, Vanita V. Genetic and environmental factors contributing to anophthalmia and microphthalmia: Current understanding and future directions. World J Clin Pediatr 2025; 14:101982. [DOI: 10.5409/wjcp.v14.i2.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/18/2025] Open
Abstract
Anophthalmia is defined as a complete absence of one eye or both the eyes, while microphthalmia represents the presence of a small eye within the orbit. The estimated birth prevalence for anophthalmia is approximately 3 per 100000 live births, and for microphthalmia, it is around 14 per 100000 live births. However, combined evidence suggests that the prevalence of these malformations could be as high as 30 per 100000 individuals. Microphthalmia is reported to occur in 3.2% to 11.2% of blind children. Anophthalmia and microphthalmia (A/M) are part of a phenotypic spectrum alongside ocular coloboma, hypothesized to share a common genetic basis. Both A/M can occur in isolation or as part of a syndrome. Their complex etiology involves chromosomal aberrations, monogenic inheritance pattern, and the contribution of environmental factors such as gestational-acquired infections, maternal vitamin A deficiency (VAD), exposure to X-rays, solvent misuse, and thalidomide exposure. A/M exhibit significant clinical and genetic heterogeneity with over 90 genes identified so far. Familial cases of A/M have a complex genetic basis, including all Mendelian modes of inheritance, i.e., autosomal dominant, recessive, and X-linked. Most cases arise sporadically due to de novo mutations. Examining gene expression during eye development and the effects of various environmental variables will help us better understand the phenotypic heterogeneity found in A/M, leading to more effective diagnosis and management strategies. The present review focuses on key genetic factors, developmental abnormalities, and environmental modifiers linked with A/M. It also emphasizes at potential research areas including multiomic methods and disease modeling with induced pluripotent stem cell technologies, which aim to create innovative treatment options.
Collapse
Affiliation(s)
- Shiwali Goyal
- Department of Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD 20852, United States
| | - Shailja Tibrewal
- Department of Pediatric Ophthalmology, Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Ria Ratna
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Vanita Vanita
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| |
Collapse
|
2
|
Zaluski J, Bassetto M, Kiser PD, Tochtrop GP. Advances and therapeutic opportunities in visual cycle modulation. Prog Retin Eye Res 2025; 106:101360. [PMID: 40280538 DOI: 10.1016/j.preteyeres.2025.101360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The visual cycle is a metabolic pathway that enables continuous vision by regenerating the 11-cis-retinal chromophore for photoreceptors opsins. Although integral to normal visual function, the flux of retinoids through this cycle can contribute to a range of retinal pathologies, including Stargardt disease, age-related macular degeneration, and diabetic retinopathy. In such conditions, intermediates and byproducts of the visual cycle, such as bisretinoid components of lipofuscin, can accumulate, concomitant with cellular damage and eventual photoreceptor loss. This has inspired efforts to modulate the visual cycle, aiming to slow or prevent the formation of these toxic intermediates and thus preserve retinal structure and function. Over the past two decades, multiple strategies to modulate the visual cycle have emerged. These include both intrinsic approaches, targeting key enzymes, retinoid-binding proteins, or receptors within the pigment epithelium or photoreceptors (e.g., RPE65, CRBP1, and rhodopsin inhibitors/antagonists) and extrinsic strategies that indirectly alter retinoid availability within the retina (e.g., RBP4 antagonists). Many of these agents have shown promise in animal models of visual cycle-associated retinal diseases, reducing pathological changes, and improving retinal survival. Several have advanced into clinical studies, although none are currently FDA-approved. Challenges remain in optimizing drug specificity and duration of action while minimizing side effects such as nyctalopia. In this review, we comprehensively examine current and emerging visual cycle modulators, discuss their medicinal chemistry, mechanisms of action, efficacy in preclinical and clinical studies, and highlight future opportunities for drug discovery aimed at safely and effectively preserving vision through modulation of this biochemical pathway.
Collapse
Affiliation(s)
- Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California- Irvine, Irvine, CA, 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA, 90822, USA; Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, CA, 92697, USA.
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
3
|
Parker LE, Papanicolaou KN, Zalesak-Kravec S, Weinberger EM, Kane MA, Foster DB. Retinoic acid signaling and metabolism in heart failure. Am J Physiol Heart Circ Physiol 2025; 328:H792-H813. [PMID: 39933792 DOI: 10.1152/ajpheart.00871.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/24/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Nearly 70 years after studies first showed that the offspring of vitamin A (retinol, ROL)-deficient rats exhibit structural cardiac defects and over 20 years since the role of vitamin A's potent bioactive metabolite hormone, all-trans retinoic acid (ATRA), was elucidated in embryonic cardiac development, the role of the vitamin A metabolites, or retinoids, in adult heart physiology and heart and vascular disease, remains poorly understood. Studies have shown that low serum levels of retinoic acid correlate with higher all-cause and cardiovascular mortality, though the relationship between circulating retinol and ATRA levels, cardiac tissue ATRA levels, and intracellular cardiac ATRA signaling in the context of heart and vascular disease has only begun to be addressed. We have recently shown that patients with idiopathic dilated cardiomyopathy show a nearly 40% decline of in situ cardiac ATRA levels, despite adequate local stores of retinol. Moreover, we and others have shown that the administration of ATRA forestalls the development of heart failure (HF) in rodent models. In this review, we summarize key facets of retinoid metabolism and signaling and discuss mechanisms by which impaired ATRA signaling contributes to several HF hallmarks including hypertrophy, contractile dysfunction, poor calcium handling, redox imbalance, and fibrosis. We highlight unresolved issues in cardiac ATRA metabolism whose pursuit will help refine therapeutic strategies aimed at restoring ATRA homeostasis.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | | | - Eva M Weinberger
- School of Medicine, Imperial College London, London, United Kingdom
| | - Maureen A Kane
- School of Pharmacy, University of Maryland, Baltimore, Maryland, United States
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Zhang X, Qi M, Fu Q. Molecular genetics of congenital heart disease. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2861-9. [PMID: 40163266 DOI: 10.1007/s11427-024-2861-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/08/2025] [Indexed: 04/02/2025]
Abstract
Congenital heart disease (CHD) is the most prevalent human birth defect and remains a leading cause of mortality in childhood. Although advancements in surgical and medical interventions have significantly reduced mortality rates among infants with critical CHDs, many survivors experience substantial cardiac and extracardiac comorbidities that affect their quality of life. The etiology of CHD is multifactorial, involving both genetic and environmental factors, yet a definitive cause remains unidentified in many cases. Recent advancements in genetic testing technologies have improved our ability to identify the genetic causes of CHD. This review presents an updated summary of the established genetic contributions to CHD, including chromosomal aberrations and mutations in genes associated with transcription factors, cardiac structural proteins, chromatin modifiers, cilia-related proteins, and cell signaling pathways. Furthermore, we discuss recent findings that support the roles of non-coding mutations and complex inheritance in the etiology of CHD.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Molecular Diagnosis for Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ming Qi
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Molecular Diagnosis for Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qihua Fu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Molecular Diagnosis for Pediatrics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Sichuan Provincial People's Hospital, Sichuan Academy of Medical Sciences, Chengdu, 610072, China.
| |
Collapse
|
5
|
Russo M, Palmeri S, Zucconi A, Vagge A, Arioni C. Management of anophthalmia, microphthalmia and coloboma in the newborn, shared care between neonatologist and ophthalmologist: a literature review. Ital J Pediatr 2025; 51:65. [PMID: 40038803 PMCID: PMC11881466 DOI: 10.1186/s13052-025-01882-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Congenital ocular anomalies significantly contribute to global disability, with 15-20% of infant blindness attributed to these anomalies. This study examined anophthalmia, microphthalmia, and coloboma (AMC) through collaborative neonatology and ophthalmology care.The global prevalence of AMC varies: anophthalmia at 0.6-4.2 per 100,000 births and microphthalmia at 2-17 per 100,000 births, with a combined prevalence of up to 30 per 100,000. The prevalence of coloboma, alone or associate with other eye defects is 2-19 per 100,000 live births. Anophthalmia and microphthalmia may present as isolated or genetic syndromes, necessitating comprehensive evaluation. AMC etiology encompasses genetic and environmental factors. Chromosomal aberrations and mutations in genes such as PAX6, SOX2, OTX2, and CHD7 are contributors. Syndromic associations, such as CHARGE (heart defect, atresia choanae, retarded growth and development, genital hypoplasia, ear anomalies/deafness) syndrome, underscore the complexity of this syndrome. Early AMC diagnosis is pivotal for timely intervention. This work provides a literature review offering insights for effective management and genetic counseling in a pediatric context.
Collapse
Affiliation(s)
- Monica Russo
- Operative Unit of Neonatology, IRCCS Ospedale Policlinico San Martino, Genoa, 16132, Italy.
| | - Serena Palmeri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Sciences (DINOGMI), University of Genoa, Genoa, Italy
| | - Alice Zucconi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Sciences (DINOGMI), University of Genoa, Genoa, Italy
| | - Aldo Vagge
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Clinica Oculistica, DINOGMI, Università di Genova, Genoa, Italy.
- Fondazione Chiossone, Genoa, Italy.
| | - Cesare Arioni
- Operative Unit of Neonatology, IRCCS Ospedale Policlinico San Martino, Genoa, 16132, Italy
| |
Collapse
|
6
|
Ge H, Di G, Song P, Han W, Chen P, Wang Y. Role of vitamin A on the ocular surface. Exp Eye Res 2025; 250:110179. [PMID: 39581361 DOI: 10.1016/j.exer.2024.110179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Vitamin A is an essential fat-soluble vitamin that cannot be endogenously synthesized by the human body. Retinoic acid (RA) is the biologically active form of vitamin A. Utilizing both nuclear and non-nuclear receptor-mediated pathways, RA plays a crucial role in regulating various biological processes, including apoptosis, differentiation, and anti-inflammatory properties within the cornea and conjunctiva. In addition, RA has been demonstrated to exert a significant influence on anti-tumor mechanisms. Disruption of RA signaling can result in corneal defects, anophthalmia, and microphthalmia. However, the beneficial effects of RA are only observed when it is administered at appropriate dosages, and higher doses have an adverse impact. Ocular abnormalities are often early indicators of a vitamin A deficiency. The lacrimal gland secretes vitamin A onto the ocular surface, where it is metabolized into RA via two sequential steps. This article provides a comprehensive overview of how vitamin A is transformed and transported from the intestine to the ocular surface, ultimately contributing to the maintenance of the normal physiological function of the ocular surface.
Collapse
Affiliation(s)
- Huanhuan Ge
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Guohu Di
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Peirong Song
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Wenshuo Han
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China; Department of Ophthalmology, Qingdao Eighth People's Hospital, Qingdao, Shandong, 266121, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Ye Wang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China.
| |
Collapse
|
7
|
Wang MH, Li GF, Zhang J. Bibliometric analysis of microphthalmos and anophthalmos over 20 years: from 2004 to 2023. Int J Ophthalmol 2024; 17:2120-2128. [PMID: 39559321 PMCID: PMC11528269 DOI: 10.18240/ijo.2024.11.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/16/2024] [Indexed: 11/20/2024] Open
Abstract
AIM To conduct a bibliometric analysis of studies on microphthalmos and anophthalmos (M/A), explore research hotspots, and provide information on future research interests in this field to benefit clinicians and researchers. METHODS Totally 751 publications related to M/A from the year 2004 to 2023 were collected from the Web of Science Core Collection database. These publications consist of both original and review articles, that are composed in English. The contributions of different countries, institutions, journals, and authors were analyzed, and network analysis was conducted by using Microsoft Excel 2021, VOSviewer, and R Studio to visualize research hotspots. RESULTS Among all publications included, the highest number of publications came from USA (218, 29.03%). China followed with 99 publications (13.18%), and England with 86 publications (11.45%). The publications from the USA had the highest frequency of citations, with 16 699 citations, and the highest H-index of 49. The American Journal of Medical Genetics Part A (43, 5.73%) published the largest number of papers, and the University of London had the most publications (41, 5.46%). The genetic and molecular mechanisms of M/A were still unclear and the clinical intervention for M/A had gained a lot of attention as an emerging area of interest. CONCLUSION Data have been gathered on the yearly count of published materials and citations, as well as the rise in publication trends, the efficiency of regions or countries, authors, journals, and organizations, along with the high-cited publications in M/A. The recent trend of research has shifted from genetic mechanisms to different clinical phenotypes and corresponding clinical interventions, which can give direction to future research.
Collapse
Affiliation(s)
- Ming-Hui Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Capital Medical University, Beijing 100730, China
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne 50937, Germany
| | - Gong-Fei Li
- Department of Neurology & Stroke, University of Tübingen, Tübingen 72076, Germany
| | - Ju Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Beijing Ophthalmology and Visual Science Key Lab, Capital Medical University, Beijing 100730, China
| |
Collapse
|
8
|
Previdi A, Jordan P, Egloff C, Coussement A, Ahmed-Eli S, Tudal L, Bienvenu T, Picone O, Dupont JM. Prenatal diagnosis of a 15q24.1 microdeletion in a fetus with cerebral and urogenital abnormalities. Clin Genet 2024; 106:537-544. [PMID: 39012202 DOI: 10.1111/cge.14592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
15q24.1 microdeletion syndrome is a recently described condition often resulting from non-allelic homologous recombination (NAHR). Typical clinical features include pre and post-natal growth retardation, facial dysmorphism, developmental delay and intellectual disability. Nonspecific urogenital, skeletal, and digit abnormalities may be present, although other congenital malformations are less frequent. Consequently, only one case was reported prenatally, complicating the genotype-phenotype correlation and the genetic counseling. We identified prenatally a second case, presenting with cerebral abnormalities including hydrocephaly, macrocephaly, cerebellum hypoplasia, vermis hypoplasia, rhombencephalosynapsis, right kidney agenesis with left kidney duplication and micropenis. Genome-wide aCGH assay allowed a diagnosis at 26 weeks of amenorrhea revealing a 1.6 Mb interstitial deletion on the long arm of chromosome 15 at 15q24.1-q24.2 (arr[GRCh37] 15q24.1q24.2(74,399,112_76,019,966)x1). A deep review of the literature was undertaken to further delineate the prenatal clinical features and the candidate genes involved in the phenotype. Cerebral malformations are typically nonspecific, but microcephaly appears to be the most frequent in postnatal cases. Our case is the first reported with a frank cerebellar involvement.
Collapse
Affiliation(s)
- Anaïk Previdi
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Pénélope Jordan
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Charles Egloff
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Aurélie Coussement
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Samira Ahmed-Eli
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Laure Tudal
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Thierry Bienvenu
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| | - Olivier Picone
- AP-HP.Nord-Université Paris Cité, Site Hôpital Louis Mourier, Service de Gynécologie Obstétrique, Colombes, France
| | - Jean-Michel Dupont
- APHP.Centre-Université Paris Cité, Site Hôpital Cochin, Service de Médecine Génomique des Maladies de Système et d'Organe, Paris, France
| |
Collapse
|
9
|
Engfer ZJ, Palczewski K. The multifaceted roles of retinoids in eye development, vision, and retinal degenerative diseases. Curr Top Dev Biol 2024; 161:235-296. [PMID: 39870435 DOI: 10.1016/bs.ctdb.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Vitamin A (all-trans-retinol; at-Rol) and its derivatives, known as retinoids, have been adopted by vertebrates to serve as visual chromophores and signaling molecules, particularly in the eye/retina. Few tissues rely on retinoids as heavily as the retina, and the study of genetically modified mouse models with deficiencies in specific retinoid-metabolizing proteins has allowed us to gain insight into the unique or redundant roles of these proteins in at-Rol uptake and storage, or their downstream roles in retinal development and function. These processes occur during embryogenesis and continue throughout life. This review delves into the role of these genes in supporting retinal function and maps the impact that genetically modified mouse models have had in studying retinoid-related genes. These models display distinct perturbations in retinoid biochemistry, physiology, and metabolic flux, mirroring human ocular diseases.
Collapse
Affiliation(s)
- Zachary J Engfer
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States.
| | - Krzysztof Palczewski
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States; Department of Chemistry, University of California Irvine, Irvine, CA, United States; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
10
|
Nakamura M, Sandell LL. Multiple roles for retinoid signaling in craniofacial development. Curr Top Dev Biol 2024; 161:33-57. [PMID: 39870438 DOI: 10.1016/bs.ctdb.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Retinoic acid (RA) signaling plays multiple essential roles in development of the head and face. Animal models with mutations in genes involved in RA signaling have enabled understanding of craniofacial morphogenic processes that are regulated by the retinoid pathway. During craniofacial morphogenesis RA signaling is active in spatially restricted domains defined by the expression of genes involved in RA production and RA breakdown. The spatial distribution of RA signaling changes with progressive development, corresponding to a multiplicity of craniofacial developmental processes that are regulated by RA. One important role of RA signaling occurs in the hindbrain. There RA contributes to specification of the anterior-posterior (AP) axis of the developing CNS and to the neural crest cells (NCC) which form the bones and nerves of the face and pharyngeal region. In the optic vesicles and frontonasal process RA orchestrates development of the midface, eyes, and nasal airway. Additional roles for RA in craniofacial development include regulation of submandibular salivary gland development and maintaining patency in the sutures of the cranial vault.
Collapse
Affiliation(s)
- Masahiro Nakamura
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States.
| |
Collapse
|
11
|
Fujikawa Y, Kato K, Unno K, Narita S, Okuno Y, Sato Y, Takefuji M, Murohara T. Dynamic upregulation of retinoic acid signal in the early postnatal murine heart promotes cardiomyocyte cell cycle exit and maturation. Sci Rep 2024; 14:20222. [PMID: 39215116 PMCID: PMC11364823 DOI: 10.1038/s41598-024-70918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The adult mammalian heart has extremely limited cardiac regenerative capacity. Most cardiomyocytes live in a state of permanent cell-cycle arrest and are unable to re-enter the cycle. Cardiomyocytes switch from cell proliferation to a maturation state during neonatal development. Although several signaling pathways are involved in this transition, the molecular mechanisms by which these inputs coordinately regulate cardiomyocyte maturation are not fully understood. Retinoic acid (RA) plays a pivotal role in development, morphogenesis, and regeneration. Despite the importance of RA signaling in embryo heart development, little is known about its function in the early postnatal period. We found that mRNA expression of aldehyde dehydrogenase 1 family member A2 (Aldh1a2), which encodes the key enzyme for synthesizing all-trans retinoic acid (ATRA) and is an important regulator for RA signaling, was transiently upregulated in neonatal mouse ventricles. Single-cell transcriptome analysis and immunohistochemistry revealed that Aldh1a2 expression was enriched in cardiac fibroblasts during the early postnatal period. Administration of ATRA inhibited cardiomyocyte proliferation in cultured neonatal rat cardiomyocytes and human cardiomyocytes. RNA-seq analysis indicated that cell proliferation-related genes were downregulated in prenatal rat ventricular cardiomyocytes treated with ATRA, while cardiomyocyte maturation-related genes were upregulated. These findings suggest that RA signaling derived from cardiac fibroblasts is one of the key regulators of cardiomyocyte proliferation and maturation during neonatal heart development.
Collapse
Affiliation(s)
- Yusuke Fujikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
| | - Kazumasa Unno
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan.
| | - Shingo Narita
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Yusuke Okuno
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Yoshitaka Sato
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
12
|
AlAbdi L, Rahbeeni Z, Maddirevula S, Helaby R, Abdulwahab F, Khan AO, Riley LG, Alhashem A, Chassaing N, Jamieson RV, Alkuraya FS. A founder variant expands the phenotype of WNT7B-related PDAC syndrome. Clin Genet 2024; 106:66-71. [PMID: 38417950 DOI: 10.1111/cge.14512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 02/15/2024] [Indexed: 03/01/2024]
Abstract
Pulmonary hypoplasia, Diaphragmatic anomalies, Anophthalmia/microphthalmia, and Cardiac defects (PDAC) syndrome is a genetically heterogeneous multiple congenital malformation syndrome. Although pathogenic variants in RARB and STRA6 are established causes of PDAC, many PDAC cases remain unsolved at the molecular level. Recently, we proposed biallelic WNT7B variants as a novel etiology based on several families with typical features of PDAC syndrome albeit with variable expressivity. Here, we report three patients from two families that share a novel founder variant in WNT7B (c.739C > T; Arg247Trp). The phenotypic expression of this variant ranges from typical PDAC features to isolated genitourinary anomalies. Similar to previously reported PDAC-associated WNT7B variants, this variant was found to significantly impair WNT7B signaling activity further corroborating its proposed pathogenicity. This report adds further evidence to WNT7B-related PDAC and expands its variable expressivity.
Collapse
Affiliation(s)
- Lama AlAbdi
- Department of Zoology, Collage of Science, King Saud University, Riyadh, Saudi Arabia
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Zuhair Rahbeeni
- Department of Medical Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sateesh Maddirevula
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rana Helaby
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Arif O Khan
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Lisa G Riley
- Rare Diseases Functional Genomics, Kids Research, The Children's Hospital at Westmead and The Children's Medical Research Institute, Sydney, New South Wales, Australia
- Specialty of Child & Adolescent Health, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Amal Alhashem
- Division of Clinical Genetic and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Genetic and Metabolic, Sehha Virtual Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Nicolas Chassaing
- Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, Purpan University Hospital, Toulouse, Midi-Pyrénées, France
- Department of Medical Genetics, Purpan University Hospital, Toulouse, Midi-Pyrénées, France
| | - Robyn V Jamieson
- Eye Genetics Research Unit, Children's Medical Research Institute, University of Sydney; The Children's Hospital at Westmead, Sydney Children's Hospitals Network; and Save Sight Institute, Sydney, New South Wales, Australia
- Specialty of Genomic Medicine, Faculty of Medicine and Health and Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Division of Clinical Genetic and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
13
|
Leung M, Steinman J, Li D, Lor A, Gruesen A, Sadah A, van Kuijk FJ, Montezuma SR, Kondkar AA, Radhakrishnan R, Lobo GP. The Logistical Backbone of Photoreceptor Cell Function: Complementary Mechanisms of Dietary Vitamin A Receptors and Rhodopsin Transporters. Int J Mol Sci 2024; 25:4278. [PMID: 38673863 PMCID: PMC11050646 DOI: 10.3390/ijms25084278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
In this review, we outline our current understanding of the mechanisms involved in the absorption, storage, and transport of dietary vitamin A to the eye, and the trafficking of rhodopsin protein to the photoreceptor outer segments, which encompasses the logistical backbone required for photoreceptor cell function. Two key mechanisms of this process are emphasized in this manuscript: ocular and systemic vitamin A membrane transporters, and rhodopsin transporters. Understanding the complementary mechanisms responsible for the generation and proper transport of the retinylidene protein to the photoreceptor outer segment will eventually shed light on the importance of genes encoded by these proteins, and their relationship on normal visual function and in the pathophysiology of retinal degenerative diseases.
Collapse
Affiliation(s)
- Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Jeremy Steinman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Dorothy Li
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Anjelynt Lor
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Ahmed Sadah
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Frederik J. van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Sandra R. Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 12271, Saudi Arabia;
| | - Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Glenn P. Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| |
Collapse
|
14
|
Nakano H, Nakano A. The role of metabolism in cardiac development. Curr Top Dev Biol 2024; 156:201-243. [PMID: 38556424 DOI: 10.1016/bs.ctdb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Metabolism is the fundamental process that sustains life. The heart, in particular, is an organ of high energy demand, and its energy substrates have been studied for more than a century. In recent years, there has been a growing interest in understanding the role of metabolism in the early differentiation of pluripotent stem cells and in cancer research. Studies have revealed that metabolic intermediates from glycolysis and the tricarboxylic acid cycle act as co-factors for intracellular signal transduction, playing crucial roles in regulating cell behaviors. Mitochondria, as the central hub of metabolism, are also under intensive investigation regarding the regulation of their dynamics. The metabolic environment of the fetus is intricately linked to the maternal metabolic status, and the impact of the mother's nutrition and metabolic health on fetal development is significant. For instance, it is well known that maternal diabetes increases the risk of cardiac and nervous system malformations in the fetus. Another notable example is the decrease in the risk of neural tube defects when pregnant women are supplemented with folic acid. These examples highlight the profound influence of the maternal metabolic environment on the fetal organ development program. Therefore, gaining insights into the metabolic environment within developing fetal organs is critical for deepening our understanding of normal organ development. This review aims to summarize recent findings that build upon the historical recognition of the environmental and metabolic factors involved in the developing embryo.
Collapse
Affiliation(s)
- Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, United States; Cardiology Division, Department of Medicine, UCLA, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, United States; Molecular Biology Institute, UCLA, Los Angeles, CA, United States; Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
15
|
Kamp JC, Neubert L, Schupp JC, Braubach P, Wrede C, Laenger F, Salditt T, Reichmann J, Welte T, Ruhparwar A, Ius F, Schwerk N, Bergmann AK, von Hardenberg S, Griese M, Rapp C, Olsson KM, Fuge J, Park DH, Hoeper MM, Jonigk DD, Knudsen L, Kuehnel MP. Multilamellated Basement Membranes in the Capillary Network of Alveolar Capillary Dysplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:180-194. [PMID: 38029923 DOI: 10.1016/j.ajpath.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/12/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
A minimal diffusion barrier is key to the pulmonary gas exchange. In alveolar capillary dysplasia (ACD), a rare genetically driven disease of early infancy, this crucial fibrovascular interface is compromised while the underlying pathophysiology is insufficiently understood. Recent in-depth analyses of vascular alterations in adult lung disease encouraged researchers to extend these studies to ACD and compare the changes of the microvasculature. Lung tissue samples of children with ACD (n = 12), adults with non-specific interstitial pneumonia (n = 12), and controls (n = 20) were studied using transmission electron microscopy, single-gene sequencing, immunostaining, exome sequencing, and broad transcriptome profiling. In ACD, pulmonary capillary basement membranes were hypertrophied, thickened, and multilamellated. Transcriptome profiling revealed increased CDH5, COL4A1, COL15A1, PTK2B, and FN1 and decreased VIT expression, confirmed by immunohistochemistry. In contrast, non-specific interstitial pneumonia samples showed a regular basement membrane architecture with preserved VIT expression but also increased COL15A1+ vessels. This study provides insight into the ultrastructure and pathophysiology of ACD. The lack of normally developed lung capillaries appeared to cause a replacement by COL15A1+ vessels, a mechanism recently described in interstitial lung disease. The VIT loss and FN1 overexpression might contribute to the unique appearance of basement membranes in ACD. Future studies are needed to explore the therapeutic potential of down-regulating the expression of FN1 and balancing VIT deficiency.
Collapse
Affiliation(s)
- Jan C Kamp
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany.
| | - Lavinia Neubert
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Jonas C Schupp
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Christoph Wrede
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany; Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Florian Laenger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Tim Salditt
- Institute of X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Jakob Reichmann
- Institute of X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Tobias Welte
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Arjang Ruhparwar
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Fabio Ius
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Nicolaus Schwerk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Clinic for Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Anke K Bergmann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | | | - Matthias Griese
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital of Ludwig Maximilian University Munich, German Center for Lung Research, Munich, Germany
| | - Christina Rapp
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital of Ludwig Maximilian University Munich, German Center for Lung Research, Munich, Germany
| | - Karen M Olsson
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Jan Fuge
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Da-Hee Park
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Marius M Hoeper
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany
| | - Danny D Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Pathology, University of Aachen, Aachen, Germany
| | - Lars Knudsen
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Mark P Kuehnel
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, German Center for Lung Research, Hannover, Germany; Institute of Pathology, University of Aachen, Aachen, Germany
| |
Collapse
|
16
|
Plaisancié J, Martinovic J, Chesneau B, Whalen S, Rodriguez D, Audebert-Bellanger S, Marzin P, Grotto S, Perthus I, Holt RJ, Bax DA, Ragge N, Chassaing N. Clinical, genetic and biochemical signatures of RBP4-related ocular malformations. J Med Genet 2023; 61:84-92. [PMID: 37586836 DOI: 10.1136/jmg-2023-109331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/16/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND The retinoic acid (RA) pathway plays a crucial role in both eye morphogenesis and the visual cycle. Individuals with monoallelic and biallelic pathogenic variants in retinol-binding protein 4 (RBP4), encoding a serum retinol-specific transporter, display variable ocular phenotypes. Although few families have been reported worldwide, recessive inherited variants appear to be associated with retinal degeneration, while individuals with dominantly inherited variants manifest ocular development anomalies, mainly microphthalmia, anophthalmia and coloboma (MAC). METHODS We report here seven new families (13 patients) with isolated and syndromic MAC harbouring heterozygous RBP4 variants, of whom we performed biochemical analyses. RESULTS For the first time, malformations that overlap the clinical spectrum of vitamin A deficiency are reported, providing a link with other RA disorders. Our data support two distinct phenotypes, depending on the nature and mode of inheritance of the variants: dominantly inherited, almost exclusively missense, associated with ocular malformations, in contrast to recessive, mainly truncating, associated with retinal degeneration. Moreover, we also confirm the skewed inheritance and impact of maternal RBP4 genotypes on phenotypical expression in dominant forms, suggesting that maternal RBP4 genetic status and content of diet during pregnancy may modify MAC occurrence and severity. Furthermore, we demonstrate that retinol-binding protein blood dosage in patients could provide a biological signature crucial for classifying RBP4 variants. Finally, we propose a novel hypothesis to explain the mechanisms underlying the observed genotype-phenotype correlations in RBP4 mutational spectrum. CONCLUSION Dominant missense variants in RBP4 are associated with MAC of incomplete penetrance with maternal inheritance through a likely dominant-negative mechanism.
Collapse
Affiliation(s)
- Julie Plaisancié
- Laboratoire National de Référence (LBMR), Génétique des anomalies malformatives de l'œil, CHU Toulouse, Toulouse, France
- Unité ToNIC Inserm 1214, CHU Toulouse, Toulouse, France
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| | - Jelena Martinovic
- Département de Génétique, Unité de Fœtopathologie, Hopital Necker-Enfants Malades, Paris, France
| | - Bertrand Chesneau
- Laboratoire National de Référence (LBMR), Génétique des anomalies malformatives de l'œil, CHU Toulouse, Toulouse, France
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| | - Sandra Whalen
- Genetique Medicale, Hopital Armand-Trousseau, Paris, France
| | - Diana Rodriguez
- Département de Génétique, Hôpitaux Universitaires Paris Ile-de-France Ouest, Paris, France
| | | | - Pauline Marzin
- Fédération de Génétique et Médecine Génomique, Service de Médecine Génomique des Maladies Rares, Necker-Enfants Malades Hospitals, Paris, France
| | - Sarah Grotto
- Maternité Port-Royal, FHU PREMA, Hôpital Cochin, Paris, France
| | - Isabelle Perthus
- Centre d'Etude des Malformations Congénitales en Auvergne, Génétique Médicale, CHU Estaing, Clermont-Ferrand, France
| | - Richard James Holt
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Dorine A Bax
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Nicola Ragge
- Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
- West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Nicolas Chassaing
- Laboratoire National de Référence (LBMR), Génétique des anomalies malformatives de l'œil, CHU Toulouse, Toulouse, France
- Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO), CHU Toulouse, Toulouse, France
| |
Collapse
|
17
|
Moon J, Zhou G, Jankowsky E, von Lintig J. Vitamin A deficiency compromises the barrier function of the retinal pigment epithelium. PNAS NEXUS 2023; 2:pgad167. [PMID: 37275262 PMCID: PMC10235913 DOI: 10.1093/pnasnexus/pgad167] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/01/2023] [Indexed: 06/07/2023]
Abstract
A major cause for childhood blindness worldwide is attributed to nutritional vitamin A deficiency. Surprisingly, the molecular basis of the ensuing retinal degeneration has not been well defined. Abundant expression of the retinoid transporter STRA6 in the retinal pigment epithelium (RPE) and homeostatic blood levels of retinol-binding protein delay vitamin A deprivation of the mouse eyes. Hence, genetic dissection of STRA6 makes mice susceptible to nutritional manipulation of ocular retinoid status. We performed RNA-seq analyses and complemented the data with tests of visual physiology, ocular morphology, and retinoid biochemistry to compare eyes with different vitamin A status. Mild ocular vitamin A deficiency decreased transcripts of photoreceptor transduction pathway-related genes and increased transcripts of oxidative stress pathways. The response was associated with impaired visual sensitivity and an accumulation of fluorescent debris in the retina. Severe vitamin A deficiency did not only impair visual perception but also decreased transcripts of genes encoding cell adhesion and cellular junction proteins. This response altered cell morphology, resulted in significant changes in transport pathways of small molecules, and compromised the barrier function of the RPE. Together, our analyses characterize the molecular events underlying nutritional blindness in a novel mouse model and indicate that breakdown of the outer blood-retinal barrier contributes to retinal degeneration and photoreceptor cell death in severe vitamin A deficiency.
Collapse
Affiliation(s)
- Jean Moon
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gao Zhou
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eckhard Jankowsky
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Johannes von Lintig
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
18
|
Wagner R, Amonkar GM, Wang W, Shui JE, Bankoti K, Tse WH, High FA, Zalieckas JM, Buchmiller TL, Zani A, Keijzer R, Donahoe PK, Lerou PH, Ai X. A Tracheal Aspirate-derived Airway Basal Cell Model Reveals a Proinflammatory Epithelial Defect in Congenital Diaphragmatic Hernia. Am J Respir Crit Care Med 2023; 207:1214-1226. [PMID: 36731066 PMCID: PMC10161756 DOI: 10.1164/rccm.202205-0953oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/02/2023] [Indexed: 02/04/2023] Open
Abstract
Rationale: Congenital diaphragmatic hernia (CDH) is characterized by incomplete closure of the diaphragm and lung hypoplasia. The pathophysiology of lung defects in CDH is poorly understood. Objectives: To establish a translational model of human airway epithelium in CDH for pathogenic investigation and therapeutic testing. Methods: We developed a robust methodology of epithelial progenitor derivation from tracheal aspirates of newborns. Basal stem cells (BSCs) from patients with CDH and preterm and term non-CDH control subjects were derived and analyzed by bulk RNA sequencing, assay for transposase accessible chromatin with sequencing, and air-liquid interface differentiation. Lung sections from fetal human CDH samples and the nitrofen rat model of CDH were subjected to histological assessment of epithelial defects. Therapeutics to restore epithelial differentiation were evaluated in human epithelial cell culture and the nitrofen rat model of CDH. Measurements and Main Results: Transcriptomic and epigenetic profiling of CDH and control BSCs reveals a proinflammatory signature that is manifested by hyperactive nuclear factor kappa B and independent of severity and hernia size. In addition, CDH BSCs exhibit defective epithelial differentiation in vitro that recapitulates epithelial phenotypes found in fetal human CDH lung samples and fetal tracheas of the nitrofen rat model of CDH. Furthermore, blockade of nuclear factor kappa B hyperactivity normalizes epithelial differentiation phenotypes of human CDH BSCs in vitro and in nitrofen rat tracheas in vivo. Conclusions: Our findings have identified an underlying proinflammatory signature and BSC differentiation defects as a potential therapeutic target for airway epithelial defects in CDH.
Collapse
Affiliation(s)
- Richard Wagner
- Division of Newborn Medicine and
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Gaurang M. Amonkar
- Division of Newborn Medicine and
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wei Wang
- Division of Newborn Medicine and
| | | | | | - Wai Hei Tse
- Departments of Surgery, Pediatrics & Child Health, Physiology & Pathophysiology, University of Manitoba and Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Frances A. High
- Division of Medical Genetics, Department of Pediatrics, and
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery and
| | - Jill M. Zalieckas
- Division of Pediatric Surgery, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Terry L. Buchmiller
- Division of Pediatric Surgery, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Augusto Zani
- Department of Pediatric Surgery, University of Toronto, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Richard Keijzer
- Departments of Surgery, Pediatrics & Child Health, Physiology & Pathophysiology, University of Manitoba and Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Patricia K. Donahoe
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
19
|
Seese SE, Muheisen S, Gath N, Gross JM, Semina EV. Identification of HSPA8 as an interacting partner of MAB21L2 and an important factor in eye development. Dev Dyn 2023; 252:510-526. [PMID: 36576422 PMCID: PMC10947772 DOI: 10.1002/dvdy.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Pathogenic variants in human MAB21L2 result in microphthalmia, anophthalmia, and coloboma. The exact molecular function of MAB21L2 is currently unknown. We conducted a series of yeast two-hybrid (Y2H) experiments to determine protein interactomes of normal human and zebrafish MAB21L2/mab21l2 as well as human disease-associated variant MAB21L2-p.(Arg51Gly) using human adult retina and zebrafish embryo libraries. RESULTS These screens identified klhl31, tnpo1, TNPO2/tnpo2, KLC2/klc2, and SPTBN1/sptbn1 as co-factors of MAB21L2/mab21l2. Several factors, including hspa8 and hspa5, were found to interact with MAB21L2-p.Arg51Gly but not wild-type MAB21L2/mab21l2 in Y2H screens. Further analyses via 1-by-1 Y2H assays, co-immunoprecipitation, and mass spectrometry revealed that both normal and variant MAB21L2 interact with HSPA5 and HSPA8. In situ hybridization detected co-expression of hspa5 and hspa8 with mab21l2 during eye development in zebrafish. Examination of zebrafish mutant hspa8hi138Tg identified reduced hspa8 expression associated with severe ocular developmental defects, including small eye, coloboma, and anterior segment dysgenesis. To investigate the effects of hspa8 deficiency on the mab21l2Arg51_Phe52del allele, corresponding zebrafish double mutants were generated and found to be more severely affected than single mutant lines. CONCLUSION This study identifies heat shock proteins as interacting partners of MAB21L2/mab21l2 and suggests a role for this interaction in vertebrate eye development.
Collapse
Affiliation(s)
- Sarah E. Seese
- Department of Pediatrics The Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sanaa Muheisen
- Department of Pediatrics The Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Natalie Gath
- University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jeffrey M. Gross
- University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Elena V. Semina
- Department of Pediatrics The Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cell Biology, Neurobiology and Anatomy, The Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Children’s of Wisconsin, Milwaukee, WI 53226, USA
- Children’s Research Institute, Medical College of Wisconsin, Children’s of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
20
|
Bouasker S, Patel N, Greenlees R, Wellesley D, Fares Taie L, Almontashiri NA, Baptista J, Alghamdi MA, Boissel S, Martinovic J, Prokudin I, Holden S, Mudhar HS, Riley LG, Nassif C, Attie-Bitach T, Miguet M, Delous M, Ernest S, Plaisancié J, Calvas P, Rozet JM, Khan AO, Hamdan FF, Jamieson RV, Alkuraya FS, Michaud JL, Chassaing N. Bi-allelic variants in WNT7B disrupt the development of multiple organs in humans. J Med Genet 2023; 60:294-300. [PMID: 35790350 DOI: 10.1136/jmedgenet-2022-108475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/11/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Pulmonary hypoplasia, Diaphragmatic anomalies, Anophthalmia/microphthalmia and Cardiac defects delineate the PDAC syndrome. We aim to identify the cause of PDAC syndrome in patients who do not carry pathogenic variants in RARB and STRA6, which have been previously associated with this disorder. METHODS We sequenced the exome of patients with unexplained PDAC syndrome and performed functional validation of candidate variants. RESULTS We identified bi-allelic variants in WNT7B in fetuses with PDAC syndrome from two unrelated families. In one family, the fetus was homozygous for the c.292C>T (p.(Arg98*)) variant whereas the fetuses from the other family were compound heterozygous for the variants c.225C>G (p.(Tyr75*)) and c.562G>A (p.(Gly188Ser)). Finally, a molecular autopsy by proxy in a consanguineous couple that lost two babies due to lung hypoplasia revealed that both parents carry the p.(Arg98*) variant. Using a WNT signalling canonical luciferase assay, we demonstrated that the identified variants are deleterious. In addition, we found that wnt7bb mutant zebrafish display a defect of the swimbladder, an air-filled organ that is a structural homolog of the mammalian lung, suggesting that the function of WNT7B has been conserved during evolution for the development of these structures. CONCLUSION Our findings indicate that defective WNT7B function underlies a form of lung hypoplasia that is associated with the PDAC syndrome, and provide evidence for involvement of the WNT-β-catenin pathway in human lung, tracheal, ocular, cardiac, and renal development.
Collapse
Affiliation(s)
- Samir Bouasker
- Research Center, University Hospital Centre Sainte-Justine, Montreal H3T 1C5, Québec, Canada
| | - Nisha Patel
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Rebecca Greenlees
- Eye Genetics Research Unit, Children's Medical Research Institute, University of Sydney; The Children's Hospital at Westmead, Sydney Children's Hospitals Network; and Save Sight Institute, Sydney, New South Wales, Australia
| | - Diana Wellesley
- Wessex Clinical Genetic Service, University Hospital Southampton, Southampton, UK
| | - Lucas Fares Taie
- Laboratory Genetics in Ophthalmology, INSERM UMR1163, Imagine Institute for Genetic Diseases, Université Paris Descartes-Sorbonne, Paris, Île-de-France, France
| | - Naif A Almontashiri
- Center for Genetics and Inherited Diseases (CGID), Taibah University, Madinah, Al Madinah, Saudi Arabia.,Research Department, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Julia Baptista
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK.,Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Malak Ali Alghamdi
- Medical Genetic Division, Pediatric Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sarah Boissel
- Research Center, University Hospital Centre Sainte-Justine, Montreal H3T 1C5, Québec, Canada
| | - Jelena Martinovic
- Unit of Fetal Pathology, APHP Hopital Antoine-Beclere, Clamart, Île-de-France, France
| | - Ivan Prokudin
- Eye Genetics Research Unit, Children's Medical Research Institute, University of Sydney; The Children's Hospital at Westmead, Sydney Children's Hospitals Network; and Save Sight Institute, Sydney, New South Wales, Australia
| | - Samantha Holden
- Department of Cellular Pathology, University Hospital Southampton, Southampton, UK
| | - Hardeep-Singh Mudhar
- National Specialist Ophthalmic Pathology Service (NSOPS), Dept of Histopathology, Royal Hallamshire Hospital, Sheffield, UK
| | - Lisa G Riley
- Rare Diseases Functional Genomics Laboratory, The Children's Hospital at Westmead, Sydney Children's Hospitals Network, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Specialty of Paediatrics and Child Health, Faculty of Medicine and Health, University of Sydney, Sidney, New South Wales, Australia
| | - Christina Nassif
- Research Center, University Hospital Centre Sainte-Justine, Montreal H3T 1C5, Québec, Canada
| | - Tania Attie-Bitach
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163, Imagine Institute for Genetic Diseases, Paris, Île-de-France, France
| | - Marguerite Miguet
- Research Center, University Hospital Centre Sainte-Justine, Montreal H3T 1C5, Québec, Canada
| | - Marion Delous
- Equipe GENDEV, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, Université Lyon 1, Université St Etienne, Lyon, Auvergne-Rhône-Alpes, France
| | - Sylvain Ernest
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163, Imagine Institute for Genetic Diseases, Paris, Île-de-France, France
| | - Julie Plaisancié
- Department of Medical Genetics, Purpan University Hospital, Toulouse, Midi-Pyrénées, France.,Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, Purpan University Hospital, Toulouse, Midi-Pyrénées, France.,INSERM U1214, ToNIC, Université Toulouse III, Toulouse, France
| | - Patrick Calvas
- Department of Medical Genetics, Purpan University Hospital, Toulouse, Midi-Pyrénées, France.,Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, Purpan University Hospital, Toulouse, Midi-Pyrénées, France
| | - Jean-Michel Rozet
- Laboratory Genetics in Ophthalmology, INSERM UMR1163, Imagine Institute for Genetic Diseases, Université Paris Descartes-Sorbonne, Paris, Île-de-France, France
| | - Arif O Khan
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, Abu Dhabi, UAE
| | - Fadi F Hamdan
- Research Center, University Hospital Centre Sainte-Justine, Montreal H3T 1C5, Québec, Canada
| | - Robyn V Jamieson
- Eye Genetics Research Unit, Children's Medical Research Institute, University of Sydney; The Children's Hospital at Westmead, Sydney Children's Hospitals Network; and Save Sight Institute, Sydney, New South Wales, Australia.,Specialty of Genomic Medicine, Faculty of Medicine and Health and Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia .,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Jacques L Michaud
- Departments of Pediatrics and Neurosciences, Université de Montréal, Montreal H3T 1J4, Québec, Canada .,Departments of Pediatrics and Neurosciences, Université de Montréal, Montreal, Québec, Canada
| | - Nicolas Chassaing
- Department of Medical Genetics, Purpan University Hospital, Toulouse, Midi-Pyrénées, France .,Centre de Référence des Affections Rares en Génétique Ophtalmologique CARGO, Site Constitutif, Purpan University Hospital, Toulouse, Midi-Pyrénées, France
| |
Collapse
|
21
|
Wang P, Wu P, Wang J, Zeng Y, Jiang Y, Wang Y, Li S, Xiao X, Zhang Q. Missense Mutations in MAB21L1: Causation of Novel Autosomal Dominant Ocular BAMD Syndrome. Invest Ophthalmol Vis Sci 2023; 64:19. [PMID: 36892533 PMCID: PMC10010443 DOI: 10.1167/iovs.64.3.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Purpose Biallelic MAB21L1 variants have been reported to cause autosomal recessive cerebellar, ocular, craniofacial, and genital syndrome (COFG), whereas only five heterozygous pathogenic variants have been suspected to cause autosomal dominant (AD) microphthalmia and aniridia in eight families. This study aimed to report an AD ocular syndrome (blepharophimosis plus anterior segment and macular dysgenesis [BAMD]) syndrome based on clinical and genetic findings from patients with monoallelic MAB21L1 pathogenic variants in our cohort and reported cases. Methods Potential pathogenic variants in MAB21L1 were detected from a large in-house exome sequencing dataset. Ocular phenotypes of the patients with potential pathogenic variants in MAB21L1 were summarized, and the genotype-phenotype correlation was analyzed through a comprehensive literature review. Results Three heterozygous missense variants in MAB21L1, predicted to be damaging, were detected in 5 unrelated families, including c.152G>T in 2, c.152G>A in 2, and c.155T>G in one. All were absent from gnomAD. The variants were de novo in two families, transmitted from affected parents to offspring in two families, and with an unknown origin in the other family, demonstrating strong evidence of AD inheritance. All patients revealed similar BAMD phenotypes, including blepharophimosis, anterior segment dysgenesis, and macular dysgenesis. Genotype-phenotype analysis suggested that patients with monoallelic MAB21L1 missense variants had only ocular anomalies (BAMD), whereas patients with biallelic variants presented both ocular and extraocular symptoms. Conclusions Heterozygous pathogenic variants in MAB21L1 account for a new AD BAMD syndrome, which is completely different from COFG caused by homozygous variants in MAB21L1. Nucleotide c.152 is likely a mutation hot spot, and the encoded residue of p.Arg51 might be critical for MAB21L1.
Collapse
Affiliation(s)
- Panfeng Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Pengsen Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Junwen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yiyan Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yi Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yingwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shiqiang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xueshan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
22
|
Radhakrishnan R, Leung M, Solanki AK, Lobo GP. Mapping of the extracellular RBP4 ligand binding domain on the RBPR2 receptor for Vitamin A transport. Front Cell Dev Biol 2023; 11:1105657. [PMID: 36910150 PMCID: PMC9992173 DOI: 10.3389/fcell.2023.1105657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
The distribution of dietary vitamin A/all-trans retinol/ROL throughout the body is critical for maintaining retinoid function in peripheral tissues and for retinoid delivery to the eye in the support of visual function. In the circulation, all-trans-retinol bound to the RBP4 protein is transported and sequestered into target tissues for long-term storage. Two membrane receptors that facilitate all-trans retinol uptake from RBP4 have been proposed. While it is well established that the membrane receptor, STRA6, binds to circulatory RBP4 for ROL transport into the eye, the second vitamin A receptor, RBPR2, which is expressed in non-ocular tissues, is less characterized. Based on the structural homology between these two RBP4 receptors, published literature, and from our recent work in Rbpr2 -/- deficient mice, we hypothesized that RBPR2 might also have high-binding affinity for RBP4 and this mechanism facilitates ROL transport. Herein, we aimed to elucidate the membrane topology and putative RBP4 binding residues on RBPR2 to understand its physiological function for retinoid homeostasis. Using in silico analysis and site-directed mutagenesis, we identified a potential RBP4 binding domain on RBPR2. We employed an in vitro cell-based system and confirmed that mutations of these residues on RBPR2 affected its binding to exogenous RBP4 and subsequently vitamin A uptake. Using Surface Plasmon Resonance assays, we analyzed both the binding affinities and kinetic parameters of wild-type RBPR2 and individual mutants affecting the RBPR2-RBP4 binding domain with its physiological ligand RBP4. These studies not only revealed a putative RBP4 binding domain on RBPR2 but also provided new structural, biochemical, and critical information on its proposed role in RBP4 binding for ROL transport and retinoid homeostasis.
Collapse
Affiliation(s)
- Rakesh Radhakrishnan
- Department of Ophthalmology, University of Minnesota, Minneapolis, MN, United States
| | - Matthias Leung
- Department of Ophthalmology, University of Minnesota, Minneapolis, MN, United States
| | - Ashish K Solanki
- Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Glenn P Lobo
- Department of Ophthalmology, University of Minnesota, Minneapolis, MN, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States.,Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
23
|
Kishi JY, Liu N, West ER, Sheng K, Jordanides JJ, Serrata M, Cepko CL, Saka SK, Yin P. Light-Seq: light-directed in situ barcoding of biomolecules in fixed cells and tissues for spatially indexed sequencing. Nat Methods 2022; 19:1393-1402. [PMID: 36216958 PMCID: PMC9636025 DOI: 10.1038/s41592-022-01604-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 08/10/2022] [Indexed: 11/21/2022]
Abstract
We present Light-Seq, an approach for multiplexed spatial indexing of intact biological samples using light-directed DNA barcoding in fixed cells and tissues followed by ex situ sequencing. Light-Seq combines spatially targeted, rapid photocrosslinking of DNA barcodes onto complementary DNAs in situ with a one-step DNA stitching reaction to create pooled, spatially indexed sequencing libraries. This light-directed barcoding enables in situ selection of multiple cell populations in intact fixed tissue samples for full-transcriptome sequencing based on location, morphology or protein stains, without cellular dissociation. Applying Light-Seq to mouse retinal sections, we recovered thousands of differentially enriched transcripts from three cellular layers and discovered biomarkers for a very rare neuronal subtype, dopaminergic amacrine cells, from only four to eight individual cells per section. Light-Seq provides an accessible workflow to combine in situ imaging and protein staining with next generation sequencing of the same cells, leaving the sample intact for further analysis post-sequencing.
Collapse
Affiliation(s)
- Jocelyn Y Kishi
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Ninning Liu
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Emma R West
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Kuanwei Sheng
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jack J Jordanides
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Matthew Serrata
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Constance L Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Sinem K Saka
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Diacou R, Nandigrami P, Fiser A, Liu W, Ashery-Padan R, Cvekl A. Cell fate decisions, transcription factors and signaling during early retinal development. Prog Retin Eye Res 2022; 91:101093. [PMID: 35817658 PMCID: PMC9669153 DOI: 10.1016/j.preteyeres.2022.101093] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Raven Diacou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Prithviraj Nandigrami
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruth Ashery-Padan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
25
|
TITF1 Screening in Human Congenital Diaphragmatic Hernia (CDH). CHILDREN 2022; 9:children9081108. [PMID: 35892611 PMCID: PMC9332008 DOI: 10.3390/children9081108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/17/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
TITF1 (Thyroid Transcription Factor-1) is a homeodomain-containing transcription factor. Previous studies showed that Titf1 null mice are characterized by failure of tracheo-oesophageal separation and impaired lung morphogenesis resulting in Pulmonary Hypoplasia (PH). In this study, we aim to evaluate the role of TITF1 in the pathogenesis of congenital diaphragmatic hernia (CDH) in humans. We investigated TITF1 expression in human trachea and lungs and performed direct mutation analysis in a CDH population. We studied 13 human fetuses at 14 to 24 weeks of gestation. Five μm sections were fixed in paraformaldehyde and incubated with anti-TITF1 primary antibody. Positive staining was visualized by biotinylated secondary antibody. We also performed TITF1 screening on genomic DNA extracted from peripheral blood of 16 patients affected by CDH and different degrees of PH, searching for mutations, insertions, and/or deletions, by sequencing the exonic regions of the gene. Histochemical studies showed positive brown staining of fetal follicular thyroid epithelium, normal fetal trachea, and normal fetal lung bronchial epithelium. Fetal esophageal wall was immunohistochemically negative. Molecular genetic analysis showed complete identity between the sequences obtained and the Wild Type (WT) form of the gene in all cases. No mutation, insertion and/or deletion was detected. Although TITF1 is expressed in the human fetal lung and has been considered to have a role in the pathogenesis of PH in CDH, the results of our study do not support the hypothesis that TITF1 mutations play a key role in the etiopathogenesis of CDH.
Collapse
|
26
|
Baker NC, Pierro JD, Taylor LW, Knudsen TB. Identifying candidate reference chemicals for in vitro testing of the retinoid pathway for predictive developmental toxicity. ALTEX 2022; 40:217–236. [PMID: 35796328 PMCID: PMC10765368 DOI: 10.14573/altex.2202231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/21/2022] [Indexed: 11/23/2022]
Abstract
Evaluating chemicals for potential in vivo toxicity based on their in vitro bioactivity profile is an important step toward animal- free testing. A compendium of reference chemicals and data describing their bioactivity on specific molecular targets, cellular pathways, and biological processes is needed to bolster confidence in the predictive value of in vitro hazard detection. Endogenous signaling by all-trans retinoic acid (ATRA) is an important pathway in developmental processes and toxicities. Employing data extraction methods and advanced literature extraction tools, we assembled a set of candidate reference chemicals with demonstrated activity on ten protein family targets in the retinoid system. The compendium was culled from Protein Data Bank, ChEMBL, ToxCast/Tox21, and the biomedical literature in PubMed. Finally, we performed a case study on one chemical in our collection, citral, an inhibitor of endogenous ATRA production, to determine whether the literature supports an adverse outcome pathway explaining the compound’s developmental toxicity initiated by disruption of the retinoid pathway. We also deliver an updated Abstract Sifter tool populated with these reference compounds and complex search terms designed to query the literature for the downstream consequences to support concordance with targeted retinoid pathway disruption.
Collapse
Affiliation(s)
| | - Jocylin D. Pierro
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Laura W. Taylor
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Thomas B. Knudsen
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| |
Collapse
|
27
|
Zenkel M, Hoja U, Gießl A, Berner D, Hohberger B, Weller JM, König L, Hübner L, Ostermann TA, Gusek-Schneider GC, Kruse FE, Pasutto F, Schlötzer-Schrehardt U. Dysregulated Retinoic Acid Signaling in the Pathogenesis of Pseudoexfoliation Syndrome. Int J Mol Sci 2022; 23:ijms23115977. [PMID: 35682657 PMCID: PMC9180992 DOI: 10.3390/ijms23115977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 01/15/2023] Open
Abstract
Pseudoexfoliation (PEX) syndrome, a stress-induced fibrotic matrix process, is the most common recognizable cause of open-angle glaucoma worldwide. The recent identification of PEX-associated gene variants uncovered the vitamin A metabolic pathway as a factor influencing the risk of disease. In this study, we analyzed the role of the retinoic acid (RA) signaling pathway in the PEX-associated matrix metabolism and evaluated its targeting as a potential candidate for an anti-fibrotic intervention. We provided evidence that decreased expression levels of RA pathway components and diminished RA signaling activity occur in an antagonistic crosstalk with TGF-β1/Smad signaling in ocular tissues and cells from PEX patients when compared with age-matched controls. Genetic and pharmacologic modes of RA pathway inhibition induced the expression and production of PEX-associated matrix components by disease-relevant cell culture models in vitro. Conversely, RA signaling pathway activation by natural and synthetic retinoids was able to suppress PEX-associated matrix production and formation of microfibrillar networks via antagonization of Smad-dependent TGF-β1 signaling. The findings indicate that deficient RA signaling in conjunction with hyperactivated TGF-β1/Smad signaling is a driver of PEX-associated fibrosis, and that restoration of RA signaling may be a promising strategy for anti-fibrotic intervention in patients with PEX syndrome and glaucoma.
Collapse
Affiliation(s)
- Matthias Zenkel
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Ursula Hoja
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Andreas Gießl
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Daniel Berner
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
- Genetikum, 89231 Neu-Ulm, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Julia M. Weller
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Loretta König
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Lisa Hübner
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Thomas A. Ostermann
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Gabriele C. Gusek-Schneider
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Friedrich E. Kruse
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
| | - Francesca Pasutto
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.Z.); (U.H.); (A.G.); (D.B.); (B.H.); (J.M.W.); (L.K.); (L.H.); (T.A.O.); (G.C.G.-S.); (F.E.K.)
- Correspondence: ; Tel.: +49-9131-8534433; Fax: +49-9131-8534631
| |
Collapse
|
28
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming increasingly common as the global economy grows and living standards improve. Timely and effective preventions and treatments for NAFLD are urgently needed. Retinol-binding protein-4 (RBP4), the protein that transports retinol through the circulation, was found to be positively related to diabetes, obesity, cardiovascular disease, and other metabolic diseases. Observational studies on the association between serum RBP4 level and the prevalence of NAFLD found contradictory results. Some of the underlying mechanisms responsible for this association have been revealed, and the possible clinical implications of treating NAFLD by targeting RBP4 have been demonstrated. Future studies should focus on the predictive value of RBP4 on NAFLD development and its potential as a therapeutic target in NAFLD.
Collapse
Affiliation(s)
- Hangkai Huang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | | |
Collapse
|
29
|
Friedmacher F, Rolle U, Puri P. Genetically Modified Mouse Models of Congenital Diaphragmatic Hernia: Opportunities and Limitations for Studying Altered Lung Development. Front Pediatr 2022; 10:867307. [PMID: 35633948 PMCID: PMC9136148 DOI: 10.3389/fped.2022.867307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common and life-threatening birth defect, characterized by an abnormal opening in the primordial diaphragm that interferes with normal lung development. As a result, CDH is accompanied by immature and hypoplastic lungs, being the leading cause of morbidity and mortality in patients with this condition. In recent decades, various animal models have contributed novel insights into the pathogenic mechanisms underlying CDH and associated pulmonary hypoplasia. In particular, the generation of genetically modified mouse models, which show both diaphragm and lung abnormalities, has resulted in the discovery of multiple genes and signaling pathways involved in the pathogenesis of CDH. This article aims to offer an up-to-date overview on CDH-implicated transcription factors, molecules regulating cell migration and signal transduction as well as components contributing to the formation of extracellular matrix, whilst also discussing the significance of these genetic models for studying altered lung development with regard to the human situation.
Collapse
Affiliation(s)
- Florian Friedmacher
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Prem Puri
- Beacon Hospital, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Steinhoff JS, Lass A, Schupp M. Retinoid Homeostasis and Beyond: How Retinol Binding Protein 4 Contributes to Health and Disease. Nutrients 2022; 14:1236. [PMID: 35334893 PMCID: PMC8951293 DOI: 10.3390/nu14061236] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Retinol binding protein 4 (RBP4) is the specific transport protein of the lipophilic vitamin A, retinol, in blood. Circulating RBP4 originates from the liver. It is secreted by hepatocytes after it has been loaded with retinol and binding to transthyretin (TTR). TTR association prevents renal filtration due to the formation of a higher molecular weight complex. In the circulation, RBP4 binds to specific membrane receptors, thereby delivering retinol to target cells, rendering liver-secreted RBP4 the major mechanism to distribute hepatic vitamin A stores to extrahepatic tissues. In particular, binding of RBP4 to 'stimulated by retinoic acid 6' (STRA6) is required to balance tissue retinoid responses in a highly homeostatic manner. Consequently, defects/mutations in RBP4 can cause a variety of conditions and diseases due to dysregulated retinoid homeostasis and cover embryonic development, vision, metabolism, and cardiovascular diseases. Aside from the effects related to retinol transport, non-canonical functions of RBP4 have also been reported. In this review, we summarize the current knowledge on the regulation and function of RBP4 in health and disease derived from murine models and human mutations.
Collapse
Affiliation(s)
- Julia S. Steinhoff
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular Metabolic Renal (CMR)-Research Center, 10115 Berlin, Germany;
| | - Achim Lass
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria;
- Field of Excellence BioHealth, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria
| | - Michael Schupp
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular Metabolic Renal (CMR)-Research Center, 10115 Berlin, Germany;
| |
Collapse
|
31
|
Scott DA, Gofin Y, Berry AM, Adams AD. Underlying genetic etiologies of congenital diaphragmatic hernia. Prenat Diagn 2022; 42:373-386. [PMID: 35037267 PMCID: PMC8924940 DOI: 10.1002/pd.6099] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/09/2022]
Abstract
Congenital diaphragmatic hernia (CDH) is often detectable prenatally. Advances in genetic testing have made it possible to obtain a molecular diagnosis in many fetuses with CDH. Here, we review the aneuploidies, copy number variants (CNVs), and single genes that have been clearly associated with CDH. We suggest that array-based CNV analysis, with or without a chromosome analysis, is the optimal test for identifying chromosomal abnormalities and CNVs in fetuses with CDH. To identify causative sequence variants, whole exome sequencing (WES) is the most comprehensive strategy currently available. Whole genome sequencing (WGS) with CNV analysis has the potential to become the most efficient and effective means of identifying an underlying diagnosis but is not yet routinely available for prenatal diagnosis. We describe how to overcome and address the diagnostic and clinical uncertainty that may remain after genetic testing, and review how a molecular diagnosis may impact recurrence risk estimations, mortality rates, and the availability and outcomes of fetal therapy. We conclude that after the prenatal detection of CDH, patients should be counseled about the possible genetic causes of the CDH, and the genetic testing modalities available to them, in accordance with generally accepted guidelines for pretest counseling in the prenatal setting.
Collapse
Affiliation(s)
- Daryl A. Scott
- Texas Children’s Hospital, Houston, TX, 77030,
USA,Department of Molecular and Human Genetics, Baylor College
of Medicine, Houston, TX, 77030, USA,Department of Molecular Physiology and Biophysics, Baylor
College of Medicine, Houston, TX, 77030, USA,Correspondence: Daryl A. Scott, R813, One Baylor
Plaza. BCM225, Houston, TX 77030, USA, Phone: +1 713-203-7242,
| | - Yoel Gofin
- Texas Children’s Hospital, Houston, TX, 77030,
USA,Department of Molecular and Human Genetics, Baylor College
of Medicine, Houston, TX, 77030, USA
| | - Aliska M. Berry
- Department of Molecular and Human Genetics, Baylor College
of Medicine, Houston, TX, 77030, USA
| | - April D. Adams
- Department of Molecular and Human Genetics, Baylor College
of Medicine, Houston, TX, 77030, USA,Department of Obstetrics and Gynecology, Division of
Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
32
|
Scott TM, Campbell IM, Hernandez-Garcia A, Lalani SR, Liu P, Shaw CA, Rosenfeld JA, Scott DA. Clinical exome sequencing data reveal high diagnostic yields for congenital diaphragmatic hernia plus (CDH+) and new phenotypic expansions involving CDH. J Med Genet 2022; 59:270-278. [PMID: 33461977 PMCID: PMC8286264 DOI: 10.1136/jmedgenet-2020-107317] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/17/2020] [Accepted: 12/26/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Congenital diaphragmatic hernia (CDH) is a life-threatening birth defect that often co-occurs with non-hernia-related anomalies (CDH+). While copy number variant (CNV) analysis is often employed as a diagnostic test for CDH+, clinical exome sequencing (ES) has not been universally adopted. METHODS We analysed a clinical database of ~12 000 test results to determine the diagnostic yields of ES in CDH+ and to identify new phenotypic expansions. RESULTS Among the 76 cases with an indication of CDH+, a molecular diagnosis was made in 28 cases for a diagnostic yield of 37% (28/76). A provisional diagnosis was made in seven other cases (9%; 7/76). Four individuals had a diagnosis of Kabuki syndrome caused by frameshift variants in KMT2D. Putatively deleterious variants in ALG12 and EP300 were each found in two individuals, supporting their role in CDH development. We also identified individuals with de novo pathogenic variants in FOXP1 and SMARCA4, and compound heterozygous pathogenic variants in BRCA2. The role of these genes in CDH development is supported by the expression of their mouse homologs in the developing diaphragm, their high CDH-specific pathogenicity scores generated using a previously validated algorithm for genome-scale knowledge synthesis and previously published case reports. CONCLUSION We conclude that ES should be ordered in cases of CDH+ when a specific diagnosis is not suspected and CNV analyses are negative. Our results also provide evidence in favour of phenotypic expansions involving CDH for genes associated with ALG12-congenital disorder of glycosylation, Rubinstein-Taybi syndrome, Fanconi anaemia, Coffin-Siris syndrome and FOXP1-related disorders.
Collapse
Affiliation(s)
- Tiana M. Scott
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, UT, 84602, USA,Texas Children’s Hospital, Houston, TX, 77030, USA
| | - Ian M. Campbell
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andres Hernandez-Garcia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Seema R. Lalani
- Texas Children’s Hospital, Houston, TX, 77030, USA,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA,Baylor Genetics, Houston, TX, 77021, USA
| | - Chad A. Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daryl A. Scott
- Texas Children’s Hospital, Houston, TX, 77030, USA,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA,Correspondence Daryl A. Scott, R813, One Baylor Plaza. BCM225, Houston, TX 77030, USA, , Phone: +1 713-203-7242
| |
Collapse
|
33
|
Chen X, Qiu T, Xiao P, Li W. Retinal toxicity of isoflucypram to zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 243:106073. [PMID: 34999466 DOI: 10.1016/j.aquatox.2021.106073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
Isoflucypram is an active succinate dehydrogenase inhibitor (SDHI) fungicide. Recent studies have demonstrated that isoflucypram is toxic to non-target aquatic organisms such as zebrafish, Danio rerio. However, current knowledge of the potential risks presented by the SDHI to non-target aquatic organism remains limited. To investigate the teratogenic effects of isoflucypram on retinogenesis, zebrafish embryos were exposed to isoflucypram (0.025, 0.25, and 2.5 μM) from the blastula stage (3 h post-fertilization, hpf) to the larval stage (96 hpf). Prolonged exposure to isoflucypram induced abnormalities in retinal development in zebrafish larvae, resulted in the expression of a microphthalmic phenotype, disrupted retinal lamination, and altered the expression levels of retinal markers (opn1sw1, opn1sw2, opn1mw1, opn1lw1, rho, atoh7, vsx1, prox1a, and sox2). Retinal cell apoptosis was also significantly higher in the isoflucypram-exposed larvae than in the control larvae. Catalase activity decreased significantly and malondialdehyde content increased markedly after exposure to isoflucypram. Thus, isoflucypram should be regarded as having retinal neurotoxicity in zebrafish.
Collapse
Affiliation(s)
- Xin Chen
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China
| | - Tiantong Qiu
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China
| | - Peng Xiao
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
34
|
Sajovic J, Meglič A, Glavač D, Markelj Š, Hawlina M, Fakin A. The Role of Vitamin A in Retinal Diseases. Int J Mol Sci 2022; 23:1014. [PMID: 35162940 PMCID: PMC8835581 DOI: 10.3390/ijms23031014] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/24/2022] Open
Abstract
Vitamin A is an essential fat-soluble vitamin that occurs in various chemical forms. It is essential for several physiological processes. Either hyper- or hypovitaminosis can be harmful. One of the most important vitamin A functions is its involvement in visual phototransduction, where it serves as the crucial part of photopigment, the first molecule in the process of transforming photons of light into electrical signals. In this process, large quantities of vitamin A in the form of 11-cis-retinal are being isomerized to all-trans-retinal and then quickly recycled back to 11-cis-retinal. Complex machinery of transporters and enzymes is involved in this process (i.e., the visual cycle). Any fault in the machinery may not only reduce the efficiency of visual detection but also cause the accumulation of toxic chemicals in the retina. This review provides a comprehensive overview of diseases that are directly or indirectly connected with vitamin A pathways in the retina. It includes the pathophysiological background and clinical presentation of each disease and summarizes the already existing therapeutic and prospective interventions.
Collapse
Affiliation(s)
- Jana Sajovic
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Andrej Meglič
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Damjan Glavač
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Špela Markelj
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Marko Hawlina
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| | - Ana Fakin
- Eye Hospital, University Medical Centre Ljubljana, Grablovičeva 46, 1000 Ljubljana, Slovenia
| |
Collapse
|
35
|
Zhang Y, Liu X, Gao H, Cui W, Zhang B, Zhao Y. Molecular and phenotypic characteristics of 15q24 microdeletion in pediatric patients with developmental disorders. Mol Cytogenet 2021; 14:57. [PMID: 34922566 PMCID: PMC8684056 DOI: 10.1186/s13039-021-00574-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022] Open
Abstract
Chromosome 15q24 microdeletion is a rare genetic disorder characterized by development delay, facial dysmorphism, congenital malformations, and occasional autism spectrum disorder (ASD). In this study, we identified five cases of 15q24 microdeletion using multiplex ligation-dependent probe amplification (MLPA) technology in a cohort of patients with developmental delay and/or intellectual disability. Two of these five cases had deletions that overlapped with the previously defined 1.1 Mb region observed in most reported cases. Two cases had smaller deletions (< 0.57 Mb) in the 15q24.1 low copy repeat (LCR) B-C region. They presented significant neurobehavioral features, suggesting that this smaller interval is critical for core phenotypes of 15q24 microdeletion syndrome. One case had minimal homozygous deletion of less than 0.11 Mb in the 15q24.1 LCR B-C region, which contained CYP1A1 (cytochrome P450 family 1 subfamily A member 1) and EDC3 (enhancer of mRNA decapping 3) genes, resulting in poor immunity, severe laryngeal stridor, and lower limbs swelling. This study provides additional evidence of 15q24 microdeletion syndrome with genetic and clinical findings. The results will be of significance to pediatricians in their daily practice.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Xiaoliang Liu
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Haiming Gao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Wanting Cui
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Bijun Zhang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
36
|
Bendixen C, Brosens E, Chung WK. Genetic Diagnostic Strategies and Counseling for Families Affected by Congenital Diaphragmatic Hernia. Eur J Pediatr Surg 2021; 31:472-481. [PMID: 34911129 DOI: 10.1055/s-0041-1740337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common and severe birth defect with variable clinical outcome and associated malformations in up to 60% of patients. Mortality and morbidity remain high despite advances in pre-, intra-, and postnatal management. We review the current literature and give an overview about the genetics of CDH to provide guidelines for clinicians with respect to genetic diagnostics and counseling for families. Until recently, the common practice was (molecular) karyotyping or chromosome microarray if the CDH diagnosis is made prenatally with a 10% diagnostic yield. Undiagnosed patients can be reflexed to trio exome/genome sequencing with an additional diagnostic yield of 10 to 20%. Even with a genetic diagnosis, there can be a range of clinical outcomes. All families with a child with CDH with or without additional malformations should be offered genetic counseling and testing in a family-based trio approach.
Collapse
Affiliation(s)
- Charlotte Bendixen
- Department of General, Visceral, Vascular and Thoracic Surgery, Unit of Pediatric Surgery, Universitätsklinikum Bonn, Bonn, Germany
| | - Erwin Brosens
- Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Wendy Kay Chung
- Department of Medicine, Columbia University Irving Medical Center, New York, United States.,Department of Pediatrics, Columbia University Irving Medical Center, New York, United States
| |
Collapse
|
37
|
Xiao P, Li W, Lu J, Liu Y, Luo Q, Zhang H. Effects of embryonic exposure to bixafen on zebrafish (Danio rerio) retinal development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:113007. [PMID: 34808508 DOI: 10.1016/j.ecoenv.2021.113007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 06/13/2023]
Abstract
Bixafen, a pyrazole-carboxamide fungicide, is a potent toxicant that may elicit multiple adverse effects in non-target organisms. However, knowledge of the mechanisms involved in developmental defects caused by bixafen in aquatic organisms remains limited. In this study, the effects of bixafen on retinal development were evaluated in embryo-larval zebrafish. We exposed zebrafish embryos to 0, 0.1, and 0.3 μM bixafen. Exposure of zebrafish embryos to bixafen caused severe retinal defects, including extreme microphthalmia and a significantly increased cell density of the ganglion cell layer (GCL). Compared with the controls, the expression levels of rod and cone photoreceptor marker genes (rho, opn1sw2, opn1mw1, opn1lw1, and opn1sw1) in the outer nuclear layer (ONL) were significantly downregulated after bixafen exposure. Furthermore, bixafen caused significantly increased expression levels in the GCL marker ath5 and decreased expression levels in the inner nuclear layer (INL) markers prox1a, vsx1, and sox2. Accordingly, we observed a significantly increased rate of cell apoptosis in the retina after bixafen exposure. Taken together, our data demonstrate that bixafen exhibits retinal developmental toxicity to zebrafish embryos/larvae, and thus, it may pose a significant environmental threat to aquatic organisms.
Collapse
Affiliation(s)
- Peng Xiao
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution of Life and Environmental Science, Wenzhou University, Wenzhou 325035, PR China
| | - Wenhua Li
- School of Biomedical Sciences, Huaqiao University, Xiamen 361021, PR China.
| | - Jinfang Lu
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yang Liu
- College of Life Sciences, Henan Normal University, Xinxiang 453007, PR China
| | - Qiulan Luo
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou 521041, PR China
| | - He Zhang
- National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution of Life and Environmental Science, Wenzhou University, Wenzhou 325035, PR China
| |
Collapse
|
38
|
Dhokia V, Macip S. A master of all trades - linking retinoids to different signalling pathways through the multi-purpose receptor STRA6. Cell Death Discov 2021; 7:358. [PMID: 34785649 PMCID: PMC8595884 DOI: 10.1038/s41420-021-00754-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Retinoids are a group of vitamin A-related chemicals that are essential to chordate mammals. They regulate a number of basic processes, including embryogenesis and vision. From ingestion to metabolism and the subsequent cellular effects, retinoid levels are tightly regulated in the organism to prevent toxicity. One component of this network, the membrane receptor STRA6, has been shown to be essential in facilitating the cellular entry and exit of retinol. However, recent data suggests that STRA6 may not function merely as a retinoid transporter but also act as a complex signalling hub in its own right, being able to affect cell fate through the integration of retinoid signalling with other key pathways, such as those involving p53, JAK/STAT, Wnt/β catenin and calcium. This may open new therapeutic strategies in diseases like cancer, where these pathways are often compromised. Here, we look at the growing evidence regarding the novel roles of STRA6 beyond its well characterized classic functions.
Collapse
Affiliation(s)
- Vinesh Dhokia
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- Mechanisms of Cancer and Aging Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain.
| |
Collapse
|
39
|
Martin Ask N, Leung M, Radhakrishnan R, Lobo GP. Vitamin A Transporters in Visual Function: A Mini Review on Membrane Receptors for Dietary Vitamin A Uptake, Storage, and Transport to the Eye. Nutrients 2021; 13:nu13113987. [PMID: 34836244 PMCID: PMC8620617 DOI: 10.3390/nu13113987] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 11/23/2022] Open
Abstract
Vitamins are essential compounds obtained through diet that are necessary for normal development and function in an organism. One of the most important vitamins for human physiology is vitamin A, a group of retinoid compounds and carotenoids, which generally function as a mediator for cell growth, differentiation, immunity, and embryonic development, as well as serving as a key component in the phototransduction cycle in the vertebrate retina. For humans, vitamin A is obtained through the diet, where provitamin A carotenoids such as β-carotene from plants or preformed vitamin A such as retinyl esters from animal sources are absorbed into the body via the small intestine and converted into all-trans retinol within the intestinal enterocytes. Specifically, once absorbed, carotenoids are cleaved by carotenoid cleavage oxygenases (CCOs), such as Beta-carotene 15,15’-monooxygenase (BCO1), to produce all-trans retinal that subsequently gets converted into all-trans retinol. CRBP2 bound retinol is then converted into retinyl esters (REs) by the enzyme lecithin retinol acyltransferase (LRAT) in the endoplasmic reticulum, which is then packaged into chylomicrons and sent into the bloodstream for storage in hepatic stellate cells in the liver or for functional use in peripheral tissues such as the retina. All-trans retinol also travels through the bloodstream bound to retinol binding protein 4 (RBP4), where it enters cells with the assistance of the transmembrane transporters, stimulated by retinoic acid 6 (STRA6) in peripheral tissues or retinol binding protein 4 receptor 2 (RBPR2) in systemic tissues (e.g., in the retina and the liver, respectively). Much is known about the intake, metabolism, storage, and function of vitamin A compounds, especially with regard to its impact on eye development and visual function in the retinoid cycle. However, there is much to learn about the role of vitamin A as a transcription factor in development and cell growth, as well as how peripheral cells signal hepatocytes to secrete all-trans retinol into the blood for peripheral cell use. This article aims to review literature regarding the major known pathways of vitamin A intake from dietary sources into hepatocytes, vitamin A excretion by hepatocytes, as well as vitamin A usage within the retinoid cycle in the RPE and retina to provide insight on future directions of novel membrane transporters for vitamin A in retinal cell physiology and visual function.
Collapse
|
40
|
Wiesinger A, Boink GJJ, Christoffels VM, Devalla HD. Retinoic acid signaling in heart development: Application in the differentiation of cardiovascular lineages from human pluripotent stem cells. Stem Cell Reports 2021; 16:2589-2606. [PMID: 34653403 PMCID: PMC8581056 DOI: 10.1016/j.stemcr.2021.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
Retinoic acid (RA) signaling plays an important role during heart development in establishing anteroposterior polarity, formation of inflow and outflow tract progenitors, and growth of the ventricular compact wall. RA is also utilized as a key ingredient in protocols designed for generating cardiac cell types from pluripotent stem cells (PSCs). This review discusses the role of RA in cardiogenesis, currently available protocols that employ RA for differentiation of various cardiovascular lineages, and plausible transcriptional mechanisms underlying this fate specification. These insights will inform further development of desired cardiac cell types from human PSCs and their application in preclinical and clinical research.
Collapse
Affiliation(s)
- Alexandra Wiesinger
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Cardiology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Young BD, Varney KM, Wilder PT, Costabile BK, Pozharski E, Cook ME, Godoy-Ruiz R, Clarke OB, Mancia F, Weber DJ. Physiologically Relevant Free Ca 2+ Ion Concentrations Regulate STRA6-Calmodulin Complex Formation via the BP2 Region of STRA6. J Mol Biol 2021; 433:167272. [PMID: 34592217 PMCID: PMC8568335 DOI: 10.1016/j.jmb.2021.167272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 11/28/2022]
Abstract
The interaction of calmodulin (CaM) with the receptor for retinol uptake, STRA6, involves an α-helix termed BP2 that is located on the intracellular side of this homodimeric transporter (Chen et al., 2016 [1]). In the absence of Ca2+, NMR data showed that a peptide derived from BP2 bound to the C-terminal lobe (C-lobe) of Mg2+-bound CaM (MgCaM). Upon titration of Ca2+ into MgCaM-BP2, NMR chemical shift perturbations (CSPs) were observed for residues in the C-lobe, including those in the EF-hand Ca2+-binding domains, EF3 and EF4 (CaKD = 60 ± 7 nM). As higher concentrations of free Ca2+ were achieved, CSPs occurred for residues in the N-terminal lobe (N-lobe) including those in EF1 and EF2 (CaKD = 1000 ± 160 nM). Thermodynamic and kinetic Ca2+ binding studies showed that BP2 addition increased the Ca2+-binding affinity of CaM and slowed its Ca2+ dissociation rates (koff) in both the C- and N-lobe EF-hand domains, respectively. These data are consistent with BP2 binding to the C-lobe of CaM at low free Ca2+ concentrations (<100 nM) like those found at resting intracellular levels. As free Ca2+ levels approach 1000 nM, which is typical inside a cell upon an intracellular Ca2+-signaling event, BP2 is shown here to interact with both the N- and C-lobes of Ca2+-loaded CaM (CaCaM-BP2). Because this structural rearrangement observed for the CaCaM-BP2 complex occurs as intracellular free Ca2+ concentrations approach those typical of a Ca2+-signaling event (CaKD = 1000 ± 160 nM), this conformational change could be relevant to vitamin A transport by full-length CaCaM-STRA6.
Collapse
Affiliation(s)
- Brianna D Young
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA
| | - Kristen M Varney
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), 9600 Gudelsky Dr., Rockville, MD 20850, USA
| | - Paul T Wilder
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), 9600 Gudelsky Dr., Rockville, MD 20850, USA
| | - Brianna K Costabile
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Edwin Pozharski
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), 9600 Gudelsky Dr., Rockville, MD 20850, USA
| | - Mary E Cook
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA
| | - Raquel Godoy-Ruiz
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), 9600 Gudelsky Dr., Rockville, MD 20850, USA
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - David J Weber
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), 9600 Gudelsky Dr., Rockville, MD 20850, USA.
| |
Collapse
|
42
|
Genetics of diaphragmatic hernia. Eur J Hum Genet 2021; 29:1729-1733. [PMID: 34621023 PMCID: PMC8632982 DOI: 10.1038/s41431-021-00972-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 01/14/2023] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a life-threatening malformation characterised by failure of diaphragmatic development with lung hypoplasia and persistent pulmonary hypertension of the newborn (PPHN). The incidence is 1:2000 corresponding to 8% of all major congenital malformations. Morbidity and mortality in affected newborns are very high and at present, there is no precise prenatal or early postnatal prognostication parameter to predict clinical outcome in CDH patients. Most cases occur sporadically, however, genetic causes have long been discussed to explain a proportion of cases. These range from aneuploidy to complex chromosomal aberrations and specific mutations often causing a complex phenotype exhibiting multiple malformations along with CDH. This review summarises the genetic variations which have been observed in syndromic and isolated cases of congenital diaphragmatic hernia.
Collapse
|
43
|
The diversity of lipocalin receptors. Biochimie 2021; 192:22-29. [PMID: 34534611 DOI: 10.1016/j.biochi.2021.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 11/23/2022]
Abstract
Lipocalins are important carriers of preferentially hydrophobic molecules, but they can also bind other ligands, like highly polar siderophores or intact proteins. Consequently, they are involved in a variety of physiological processes in many species. Since lipocalins are mainly extracellular proteins, they have to interact with cell receptors to exert their biological effects. In contrast to the large number of lipocalins identified in the last years, the number of receptors known is still limited. Nevertheless, some novel findings concerning the molecules involved in cellular uptake or signaling effects of lipocalins have been made recently. This review presents a detailed overview of the receptors identified so far. The methods used for isolation or identification are described and structural as well as functional information on these proteins is presented essentially in chronological order of their initial discovery.
Collapse
|
44
|
Saini A, Almasarweh S, Acosta S, Jayakar P, Janvier M, Wong TC, Salyakina D, Sasaki J. Syndromic Microphthalmia 9: Role of rapid genome sequencing and novel mutations in STRA6 gene. PROGRESS IN PEDIATRIC CARDIOLOGY 2021. [DOI: 10.1016/j.ppedcard.2021.101443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Berner D, Hoja U, Zenkel M, Ross JJ, Uebe S, Paoli D, Frezzotti P, Rautenbach RM, Ziskind A, Williams SE, Carmichael TR, Ramsay M, Topouzis F, Chatzikyriakidou A, Lambropoulos A, Sundaresan P, Ayub H, Akhtar F, Qamar R, Zenteno JC, Cruz-Aguilar M, Astakhov YS, Dubina M, Wiggs J, Ozaki M, Kruse FE, Aung T, Reis A, Khor CC, Pasutto F, Schlötzer-Schrehardt U. The protective variant rs7173049 at LOXL1 locus impacts on retinoic acid signaling pathway in pseudoexfoliation syndrome. Hum Mol Genet 2021; 28:2531-2548. [PMID: 30986821 PMCID: PMC6644155 DOI: 10.1093/hmg/ddz075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
LOXL1 (lysyl oxidase-like 1) has been identified as the major effect locus in pseudoexfoliation (PEX) syndrome, a fibrotic disorder of the extracellular matrix and frequent cause of chronic open-angle glaucoma. However, all known PEX-associated common variants show allele effect reversal in populations of different ancestry, casting doubt on their biological significance. Based on extensive LOXL1 deep sequencing, we report here the identification of a common non-coding sequence variant, rs7173049A>G, located downstream of LOXL1, consistently associated with a decrease in PEX risk (odds ratio, OR = 0.63; P = 6.33 × 10−31) in nine different ethnic populations. We provide experimental evidence for a functional enhancer-like regulatory activity of the genomic region surrounding rs7173049 influencing expression levels of ISLR2 (immunoglobulin superfamily containing leucine-rich repeat protein 2) and STRA6 [stimulated by retinoic acid (RA) receptor 6], apparently mediated by allele-specific binding of the transcription factor thyroid hormone receptor beta. We further show that the protective rs7173049-G allele correlates with increased tissue expression levels of ISLR2 and STRA6 and that both genes are significantly downregulated in tissues of PEX patients together with other key components of the STRA6 receptor-driven RA signaling pathway. siRNA-mediated downregulation of RA signaling induces upregulation of LOXL1 and PEX-associated matrix genes in PEX-relevant cell types. These data indicate that dysregulation of STRA6 and impaired retinoid metabolism are involved in the pathophysiology of PEX syndrome and that the variant rs7173049-G, which represents the first common variant at the broad LOXL1 locus without allele effect reversal, mediates a protective effect through upregulation of STRA6 in ocular tissues.
Collapse
Affiliation(s)
- Daniel Berner
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Hoja
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - James Julian Ross
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Steffen Uebe
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Paoli
- Department of Ophthalmology, Monfalcone Hospital, Gorizia, Italy
| | - Paolo Frezzotti
- Ophthalmology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Robyn M Rautenbach
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Ari Ziskind
- Division of Ophthalmology, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Susan E Williams
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Trevor R Carmichael
- Division of Ophthalmology, University of the Witwatersrand, Johannesburg, South Africa
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fotis Topouzis
- Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anthi Chatzikyriakidou
- Department of Biology and Genetics, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Lambropoulos
- Department of Biology and Genetics, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Periasamy Sundaresan
- Dr. G.Venkataswamy Eye Research Institute, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, India
| | - Humaira Ayub
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Abbottabad, Pakistan
| | - Farah Akhtar
- Pakistan Institute of Ophthalmology, Al-Shifa Trust Eye Hospital, Rawalpindi, Pakistan
| | - Raheel Qamar
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Juan C Zenteno
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico.,Biochemistry Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Marisa Cruz-Aguilar
- Genetics Department, Institute of Ophthalmology 'Conde de Valenciana', Mexico City, Mexico
| | - Yury S Astakhov
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St Petersburg, Russia
| | - Michael Dubina
- Department of Ophthalmology, Pavlov First Saint Petersburg State Medical University, St Petersburg, Russia.,St Petersburg Academic University, St Petersburg, Russia
| | - Janey Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Mineo Ozaki
- Ozaki Eye Hospital, Hyuga, Miyazaki, Japan.,Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Friedrich E Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tin Aung
- Singapore Eye Research Institute, Singapore.,Singapore National Eye Center, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Chiea Chuen Khor
- Singapore Eye Research Institute, Singapore.,Genome Institute of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Francesca Pasutto
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
46
|
Zheng J, Zhu S, Xu H, Li J, Tang H, Zhou Y, Huang Z, Liu G. miR-363-3p inhibits rat lung alveolar type II cell proliferation by downregulating STRA6 expression and induces cell apoptosis via cellular oxidative stress and G1-phase cell cycle arrest. Transl Pediatr 2021; 10:2095-2105. [PMID: 34584880 PMCID: PMC8429880 DOI: 10.21037/tp-21-303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND miR-363-3p, the retinoid signaling pathway (RSP), and its associated membrane receptor, stimulated by retinoic acid 6 (STRA6), participate in lung development. We hypothesize that miR-363-3p is involved in lung cell proliferation and apoptosis by regulating the expression of STRA6, and this study was designed to investigate the effect of changes in the expressions of miR-363-3p and the STRA6 gene on the proliferation and apoptosis of rat alveolar type II cells. METHODS To confirm our hypothesis, we used: a dual-luciferase reporter assay; cell culture and transfection; real-time quantitative polymerase chain reaction (PCR); Western blotting; a cell proliferation assay and flow cytometry analysis of the cell cycle, cell apoptosis, oxidative stress level, and mitochondrial membrane potential. RESULTS Our results showed that STRA6 is a target gene for miR-363-3p, and when the expression of miR-363-3p increased, the relative messenger RNA (mRNA) expression of STRA6 decreased, which caused a decrease in STRA6 protein synthesis and subsequent inhibition of rat lung alveolar type II cell proliferation. In contrast, inhibiting the expression of miR-363-3p promoted the proliferation of these cells. This study also found that an increased expression of miR-363-3p induced rat lung alveolar type II cell apoptosis led to an increase in the oxidative stress level, decreased mitochondrial membrane potential, and an inducement of G1-phase cell cycle arrest. CONCLUSIONS In conclusion, miR-363-3p is associated with lung cell proliferation and apoptosis, while miR-363-3p inhibits rat lung alveolar type II cell proliferation by downregulating the expression of STRA6 and induces cell apoptosis by increasing cellular oxidative stress and G1-phase cell cycle arrest.
Collapse
Affiliation(s)
- Jintao Zheng
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Shibo Zhu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huiyu Xu
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Jiequan Li
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Huajian Tang
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Yanfen Zhou
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Zhaomei Huang
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Guoqing Liu
- Department of Neonatal and Pediatric Surgery, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China.,Women and Children Medical Research Center Affiliated to Foshan Institute of Fetal Medicine, Foshan, China
| |
Collapse
|
47
|
Grand K, Skraban CM, Cohen JL, Dowsett L, Mazzola S, Tarpinian J, Bedoukian E, Nesbitt A, Denenberg B, Lulis L, Santani A, Zackai EH, Deardorff MA. Nonlethal presentations of CYP26B1-related skeletal anomalies and multiple synostoses syndrome. Am J Med Genet A 2021; 185:2766-2775. [PMID: 34160123 DOI: 10.1002/ajmg.a.62387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/04/2021] [Accepted: 05/22/2021] [Indexed: 11/10/2022]
Abstract
Retinoic acid exposures as well as defects in the retinoic acid-degrading enzyme CYP26B1 have teratogenic effects on both limb and craniofacial skeleton. An initial report of four individuals described a syndrome of fetal and infantile lethality with craniosynostosis and skeletal anomalies caused by homozygous pathogenic missense variants in CYP26B1. In contrast, a 22-year-old female was reported with a homozygous missense pathogenic variant in CYP26B1 with complex multisuture craniosynostosis and intellectual disability, suggesting that in some cases, biallelic pathogenic variants of CYP26B1 may be compatible with life. Here we describe four additional living individuals from two families with compound heterozygous pathogenic missense variants in CYP26B1. Structural assessment of these additional missense variants places them further from the catalytic site and supports a model consistent with milder nonlethal disease. In addition to previously reported findings of multisuture craniosynostosis, conductive hearing loss, joint contractures, long slender fingers, camptodactly, broad fingertips, and developmental delay/intellectual disability, skeletal imaging in our cases also revealed gracile long bones, gracile ribs, radioulnar synostosis, and carpal and/or tarsal fusions. These individuals broaden the phenotypic range of biallelic pathogenic variants in CYPB26B1 and most significantly clarify that mortality can range from perinatal lethality to survival into adulthood.
Collapse
Affiliation(s)
- Katheryn Grand
- Division of Medical Genetics, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Cara M Skraban
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jennifer L Cohen
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Leah Dowsett
- Kapi'olani Medical Center, Honolulu, Hawai'i, USA.,Department of Pediatrics, University of Hawai'i John A. Burns School of Medicine, Honolulu, Hawai'i, USA
| | - Sarah Mazzola
- Division of Pediatric Genetics, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Emma Bedoukian
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Addie Nesbitt
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Beth Denenberg
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lauren Lulis
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Elaine H Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Matthew A Deardorff
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
48
|
Cannata G, Caporilli C, Grassi F, Perrone S, Esposito S. Management of Congenital Diaphragmatic Hernia (CDH): Role of Molecular Genetics. Int J Mol Sci 2021; 22:ijms22126353. [PMID: 34198563 PMCID: PMC8231903 DOI: 10.3390/ijms22126353] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common major life-threatening birth defect that results in significant mortality and morbidity depending primarily on lung hypoplasia, persistent pulmonary hypertension, and cardiac dysfunction. Despite its clinical relevance, CDH multifactorial etiology is still not completely understood. We reviewed current knowledge on normal diaphragm development and summarized genetic mutations and related pathways as well as cellular mechanisms involved in CDH. Our literature analysis showed that the discovery of harmful de novo variants in the fetus could constitute an important tool for the medical team during pregnancy, counselling, and childbirth. A better insight into the mechanisms regulating diaphragm development and genetic causes leading to CDH appeared essential to the development of new therapeutic strategies and evidence-based genetic counselling to parents. Integrated sequencing, development, and bioinformatics strategies could direct future functional studies on CDH; could be applied to cohorts and consortia for CDH and other birth defects; and could pave the way for potential therapies by providing molecular targets for drug discovery.
Collapse
Affiliation(s)
- Giulia Cannata
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.C.); (C.C.); (F.G.)
| | - Chiara Caporilli
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.C.); (C.C.); (F.G.)
| | - Federica Grassi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.C.); (C.C.); (F.G.)
| | - Serafina Perrone
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy; (G.C.); (C.C.); (F.G.)
- Correspondence: ; Tel.: +39-0521-7047
| |
Collapse
|
49
|
Stefanovic S, Etchevers HC, Zaffran S. Outflow Tract Formation-Embryonic Origins of Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8040042. [PMID: 33918884 PMCID: PMC8069607 DOI: 10.3390/jcdd8040042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022] Open
Abstract
Anomalies in the cardiac outflow tract (OFT) are among the most frequent congenital heart defects (CHDs). During embryogenesis, the cardiac OFT is a dynamic structure at the arterial pole of the heart. Heart tube elongation occurs by addition of cells from pharyngeal, splanchnic mesoderm to both ends. These progenitor cells, termed the second heart field (SHF), were first identified twenty years ago as essential to the growth of the forming heart tube and major contributors to the OFT. Perturbation of SHF development results in common forms of CHDs, including anomalies of the great arteries. OFT development also depends on paracrine interactions between multiple cell types, including myocardial, endocardial and neural crest lineages. In this publication, dedicated to Professor Andriana Gittenberger-De Groot and her contributions to the field of cardiac development and CHDs, we review some of her pioneering studies of OFT development with particular interest in the diverse origins of the many cell types that contribute to the OFT. We also discuss the clinical implications of selected key findings for our understanding of the etiology of CHDs and particularly OFT malformations.
Collapse
|
50
|
Steinhoff JS, Lass A, Schupp M. Biological Functions of RBP4 and Its Relevance for Human Diseases. Front Physiol 2021; 12:659977. [PMID: 33790810 PMCID: PMC8006376 DOI: 10.3389/fphys.2021.659977] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Retinol binding protein 4 (RBP4) is a member of the lipocalin family and the major transport protein of the hydrophobic molecule retinol, also known as vitamin A, in the circulation. Expression of RBP4 is highest in the liver, where most of the body’s vitamin A reserves are stored as retinyl esters. For the mobilization of vitamin A from the liver, retinyl esters are hydrolyzed to retinol, which then binds to RBP4 in the hepatocyte. After associating with transthyretin (TTR), the retinol/RBP4/TTR complex is released into the bloodstream and delivers retinol to tissues via binding to specific membrane receptors. So far, two distinct RBP4 receptors have been identified that mediate the uptake of retinol across the cell membrane and, under specific conditions, bi-directional retinol transport. Although most of RBP4’s actions depend on its role in retinoid homeostasis, functions independent of retinol transport have been described. In this review, we summarize and discuss the recent findings on the structure, regulation, and functions of RBP4 and lay out the biological relevance of this lipocalin for human diseases.
Collapse
Affiliation(s)
- Julia S Steinhoff
- Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Achim Lass
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Michael Schupp
- Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|